Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Artif Organs ; 38(2): 135-41, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23889507

ABSTRACT

Elevated lipoprotein(a) (Lp(a)) is known as an independent risk factor for atherosclerosis and cardiovascular events. Regular lipid apheresis decreases elevated Lp(a) concentrations. However, there is a lack of reliable data regarding the effect of lipid apheresis on cardiovascular endpoints. To assess the effects of apheresis, we compared the occurrence of cardiovascular events in 37 patients treated regularly with lipid apheresis at the time periods of preinitiation of apheresis and during apheresis treatment. A retrospective analysis of 37 patients (35 men and two women; aged 58 years ± 11 [mean ± standard deviation]; body mass index 26 kg/m(2) ± 3; low-density lipoprotein (LDL)-cholesterol before apheresis 84 mg/dL ± 21; Lp(a) before apheresis 112 mg/dL ± 34) treated regularly with lipid apheresis was performed. Patients' medical records were screened for cardiovascular events at the preapheresis and during apheresis periods. Apheresis led to a significant reduction of lipid levels (LDL cholesterol -60%; Lp(a) -68%) measured after apheresis. The event-free survival rate after 1 year in the preapheresis period was 38% (22-54%, 95% confidence interval [CI]) vs. 75% (61-89%, 95% CI) in the during-apheresis period with a statistically significant difference (P < 0.0001). Apheresis seems to lower the progression of atherosclerosis leading to a reduced number of cardiovascular events in hyperlipoproteinemia(a). Because prospective and controlled trials are lacking, the therapeutic effectiveness of lipid apheresis can only be estimated.


Subject(s)
Blood Component Removal/methods , Cardiovascular Diseases/prevention & control , Lipoprotein(a)/blood , Aged , Cardiovascular Diseases/blood , Disease-Free Survival , Female , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Treatment Outcome
2.
Mol Ther Nucleic Acids ; 2: e131, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24193032

ABSTRACT

We recently demonstrated tumor-selective iodide uptake and therapeutic efficacy of combined radiovirotherapy after systemic delivery of the theranostic sodium iodide symporter (NIS) gene using a dendrimer-coated adenovirus. To further improve shielding and targeting we physically coated replication-selective adenoviruses carrying the hNIS gene with a conjugate consisting of cationic poly(amidoamine) (PAMAM) dendrimer linked to the peptidic, epidermal growth factor receptor (EGFR)-specific ligand GE11. In vitro experiments demonstrated coxsackie-adenovirus receptor-independent but EGFR-specific transduction efficiency. Systemic injection of the uncoated adenovirus in a liver cancer xenograft mouse model led to high levels of NIS expression in the liver due to hepatic sequestration, which were significantly reduced after coating as demonstrated by (123)I-scintigraphy. Reduction of adenovirus liver pooling resulted in decreased hepatotoxicity and increased transduction efficiency in peripheral xenograft tumors. (124)I-PET-imaging confirmed EGFR-specificity by significantly lower tumoral radioiodine accumulation after pretreatment with the EGFR-specific antibody cetuximab. A significantly enhanced oncolytic effect was observed following systemic application of dendrimer-coated adenovirus that was further increased by additional treatment with a therapeutic dose of (131)I. These results demonstrate restricted virus tropism and tumor-selective retargeting after systemic application of coated, EGFR-targeted adenoviruses therefore representing a promising strategy for improved systemic adenoviral NIS gene therapy.Molecular Therapy-Nucleic Acids (2013) 2, e131; doi:10.1038/mtna.2013.58; published online 5 November 2013.

3.
J Nucl Med ; 54(8): 1450-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23843567

ABSTRACT

UNLABELLED: Currently, major limitations for the clinical application of adenovirus-mediated gene therapy are high prevalence of neutralizing antibodies, widespread expression of the coxsackie-adenovirus receptor (CAR), and adenovirus sequestration by the liver. In the current study, we used the sodium iodide symporter (NIS) as a theranostic gene to investigate whether coating of adenovirus with synthetic dendrimers could be useful to overcome these hurdles in order to develop adenoviral vectors for combination of systemic oncolytic virotherapy and NIS-mediated radiotherapy. METHODS: We coated replication-deficient (Ad5-CMV/NIS) (CMV is cytomegalovirus) and replication-selective (Ad5-E1/AFP-E3/NIS) adenovirus serotype 5 carrying the hNIS gene with poly(amidoamine) dendrimers generation 5 (PAMAM-G5) in order to investigate transduction efficacy and altered tropism of these coated virus particles by (123)I scintigraphy and to evaluate their therapeutic potential for systemic radiovirotherapy in a liver cancer xenograft mouse model. RESULTS: After dendrimer coating, Ad5-CMV/NIS demonstrated partial protection from neutralizing antibodies and enhanced transduction efficacy in CAR-negative cells in vitro. In vivo (123)I scintigraphy of nude mice revealed significantly reduced levels of hepatic transgene expression after intravenous injection of dendrimer-coated Ad5-CMV/NIS (dcAd5-CMV/NIS). Evasion from liver accumulation resulted in significantly reduced liver toxicity and increased transduction efficiency of dcAd5-CMV/NIS in hepatoma xenografts. After PAMAM-G5 coating of the replication-selective Ad5-E1/AFP-E3/NIS, a significantly enhanced oncolytic effect was observed after intravenous application (virotherapy) that was further increased by additional treatment with a therapeutic dose of (131)I (radiovirotherapy) and was associated with markedly improved survival. CONCLUSION: These results demonstrate efficient liver detargeting and tumor retargeting of adenoviral vectors after coating with synthetic dendrimers, thereby representing a promising innovative strategy for systemic NIS gene therapy. Moreover, our study-based on the function of NIS as a theranostic gene allowing the noninvasive imaging of NIS expression by (123)I scintigraphy-provides detailed characterization of in vivo vector biodistribution and localization, level, and duration of transgene expression, essential prerequisites for exact planning and monitoring of clinical gene therapy trials that aim to individualize the NIS gene therapy concept.


Subject(s)
Adenoviridae/genetics , Dendrimers/metabolism , Liver Neoplasms/therapy , Liver Neoplasms/virology , Oncolytic Virotherapy/methods , Radiotherapy, Image-Guided/methods , Symporters/genetics , Adenoviridae/metabolism , Adenoviridae/physiology , Animals , Cell Line, Tumor , Humans , Iodine Radioisotopes/therapeutic use , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/radiotherapy , Mice , Radionuclide Imaging , Transduction, Genetic
4.
Hum Gene Ther ; 24(3): 306-16, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23402366

ABSTRACT

The tumor-homing property of mesenchymal stem cells (MSC) has lead to their use as delivery vehicles for therapeutic genes. The application of the sodium iodide symporter (NIS) as therapy gene allows noninvasive imaging of functional transgene expression by (123)I-scintigraphy or PET-imaging, as well as therapeutic application of (131)I or (188)Re. Based on the critical role of the chemokine RANTES (regulated on activation, normal T-cell expressed and presumably secreted)/CCL5 secreted by MSCs in the course of tumor stroma recruitment, use of the RANTES/CCL5 promoter should allow tumor stroma-targeted expression of NIS after MSC-mediated delivery. Using a human hepatocellular cancer (HCC) xenograft mouse model (Huh7), we investigated distribution and tumor recruitment of RANTES-NIS-engineered MSCs after systemic injection by gamma camera imaging. (123)I-scintigraphy revealed active MSC recruitment and CCL5 promoter activation in the tumor stroma of Huh7 xenografts (6.5% ID/g (123)I, biological half-life: 3.7 hr, tumor-absorbed dose: 44.3 mGy/MBq). In comparison, 7% ID/g (188)Re was accumulated in tumors with a biological half-life of 4.1 hr (tumor-absorbed dose: 128.7 mGy/MBq). Administration of a therapeutic dose of (131)I or (188)Re (55.5 MBq) in RANTES-NIS-MSC-treated mice resulted in a significant delay in tumor growth and improved survival without significant differences between (131)I and (188)Re. These data demonstrate successful stromal targeting of NIS in HCC tumors by selective recruitment of NIS-expressing MSCs and by use of the RANTES/CCL5 promoter. The resulting tumor-selective radionuclide accumulation was high enough for a therapeutic effect of (131)I and (188)Re opening the exciting prospect of NIS-mediated radionuclide therapy of metastatic cancer using genetically engineered MSCs as gene delivery vehicles.


Subject(s)
Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/diagnosis , Liver Neoplasms/therapy , Mesenchymal Stem Cells/metabolism , Symporters/metabolism , Animals , Cell Line, Tumor , Cell- and Tissue-Based Therapy , Disease Models, Animal , Female , Gene Expression , Gene Transfer Techniques , Humans , Iodine Radioisotopes/metabolism , Mice , Positron-Emission Tomography , RNA, Messenger/genetics , Symporters/genetics , Tissue Distribution , Xenograft Model Antitumor Assays
5.
Hum Gene Ther ; 22(12): 1563-74, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21851208

ABSTRACT

We reported the induction of tumor-selective iodide uptake and therapeutic efficacy of (131)I in a hepatocellular carcinoma (HCC) xenograft mouse model, using novel polyplexes based on linear polyethylenimine (LPEI), shielded by polyethylene glycol (PEG), and coupled with the epidermal growth factor receptor-specific peptide GE11 (LPEI-PEG-GE11). The aim of the current study in the same HCC model was to evaluate the potential of biodegradable nanoparticle vectors based on pseudodendritic oligoamines (G2-HD-OEI) for systemic sodium iodide symporter (NIS) gene delivery and to compare efficiency and tumor specificity with LPEI-PEG-GE11. Transfection of HCC cells with NIS cDNA, using G2-HD-OEI, resulted in a 44-fold increase in iodide uptake in vitro as compared with a 22-fold increase using LPEI-PEG-GE11. After intravenous application of G2-HD-OEI/NIS HCC tumors accumulated 6-11% ID/g (123)I (percentage of the injected dose per gram tumor tissue) with an effective half-life of 10 hr (tumor-absorbed dose, 281 mGy/MBq) as measured by (123)I scintigraphic gamma camera or single-photon emission computed tomography computed tomography (SPECT CT) imaging, as compared with 6.5-9% ID/g with an effective half-life of only 6 hr (tumor-absorbed dose, 47 mGy/MBq) for LPEI-PEG-GE11. After only two cycles of G2-HD-OEI/NIS/(131)I application, a significant delay in tumor growth was observed with markedly improved survival. A similar degree of therapeutic efficacy had been observed after four cycles of LPEI-PEG-GE11/(131)I. These results clearly demonstrate that biodegradable nanoparticles based on OEI-grafted oligoamines show increased efficiency for systemic NIS gene transfer in an HCC model with similar tumor selectivity as compared with LPEI-PEG-GE11, and therefore represent a promising strategy for NIS-mediated radioiodine therapy of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Genetic Therapy , Iodine Radioisotopes/therapeutic use , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Symporters/genetics , Animals , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Proliferation , Combined Modality Therapy , Drug Delivery Systems , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Humans , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Iodine Radioisotopes/pharmacokinetics , Liver Neoplasms/genetics , Mice , Mice, Nude , Multimodal Imaging , Polyethylene Glycols/administration & dosage , Polyethyleneimine/administration & dosage , Positron-Emission Tomography , RNA, Messenger/genetics , Radiotherapy , Real-Time Polymerase Chain Reaction , Tomography, X-Ray Computed , Tumor Cells, Cultured
6.
Mol Ther ; 19(9): 1704-13, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21587211

ABSTRACT

Due to its dual role as reporter and therapy gene, the sodium iodide symporter (NIS) allows noninvasive imaging of functional NIS expression by (123)I-scintigraphy or (124)I-PET imaging before the application of a therapeutic dose of (131)I. NIS expression provides a novel mechanism for the evaluation of mesenchymal stem cells (MSCs) as gene delivery vehicles for tumor therapy. In the current study, we stably transfected bone marrow-derived CD34(-) MSCs with NIS cDNA (NIS-MSC), which revealed high levels of functional NIS protein expression. In mixed populations of NIS-MSCs and hepatocellular cancer (HCC) cells, clonogenic assays showed a 55% reduction of HCC cell survival after (131)I application. We then investigated body distribution of NIS-MSCs by (123)I-scintigraphy and (124)I-PET imaging following intravenous (i.v.) injection of NIS-MSCs in a HCC xenograft mouse model demonstrating active MSC recruitment into the tumor stroma which was confirmed by immunohistochemistry and ex vivo γ-counter analysis. Three cycles of systemic MSC-mediated NIS gene delivery followed by (131)I application resulted in a significant delay in tumor growth. Our results demonstrate tumor-specific accumulation and therapeutic efficacy of radioiodine after MSC-mediated NIS gene delivery in HCC tumors, opening the prospect of NIS-mediated radionuclide therapy of metastatic cancer using MSCs as gene delivery vehicles.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Gene Transfer Techniques , Liver Neoplasms/radiotherapy , Mesenchymal Stem Cells/pathology , Symporters/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Blotting, Western , Carcinoma, Hepatocellular/genetics , Cell Survival , Female , Fluorescent Antibody Technique , Genetic Therapy/methods , Hep G2 Cells , Humans , Iodine Radioisotopes/pharmacokinetics , Liver Neoplasms/genetics , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Symporters/metabolism , Transfection/methods
7.
Hum Gene Ther ; 22(11): 1403-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21488714

ABSTRACT

We reported the therapeutic efficacy of (131)I in hepatocellular carcinoma (HCC) cells stably expressing the sodium iodide symporter (NIS) under the control of the tumor-specific α-fetoprotein (AFP) promoter. In the current study we investigated the efficacy of adenovirus-mediated in vivo NIS gene transfer followed by (131)I and (188)Re administration for the treatment of HCC xenografts. We used a replication-deficient adenovirus carrying the human NIS gene linked to the mouse AFP promoter (Ad5-AFP-NIS) for in vitro and in vivo NIS gene transfer. Functional NIS expression was confirmed by in vivo γ-camera imaging, followed by analysis of NIS protein and mRNA expression. Human HCC (HepG2) cells infected with Ad5-AFP-NIS concentrated 50% of the applied activity of (125)I, which was sufficiently high for a therapeutic effect in an in vitro clonogenic assay. Four days after intratumoral injection of Ad5-AFP-NIS (3×10(9) plaque-forming units) HepG2 xenografts accumulated 14.5% injected dose (ID)/g (123)I with an effective half-life of 13 hr (tumor-absorbed dose, 318 mGy/MBq (131)I). In comparison, 9.2% ID/g (188)Re was accumulated in tumors with an effective half-life of 12.8 hr (tumor-absorbed dose, 545 mGy/MBq). After adenovirus-mediated NIS gene transfer in HepG2 xenografts administration of a therapeutic dose of (131)I or (188)Re (55.5 MBq) resulted in a significant delay in tumor growth and improved survival without a significant difference between (188)Re and (131)I. In conclusion, a therapeutic effect of (131)I and (188)Re was demonstrated in HepG2 xenografts after tumor-specific adenovirus-mediated in vivo NIS gene transfer.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Iodine Radioisotopes/administration & dosage , Liver Neoplasms/therapy , Radioisotopes/administration & dosage , Rhenium/administration & dosage , Symporters/genetics , Adenoviridae/genetics , Animals , Cell Line, Tumor , Genetic Therapy , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , Promoter Regions, Genetic , RNA, Messenger/metabolism , Transfection , Transplantation, Heterologous
8.
Mol Ther ; 19(4): 676-85, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21245850

ABSTRACT

We recently demonstrated tumor-selective iodide uptake and therapeutic efficacy of radioiodine in neuroblastoma tumors after systemic nonviral polyplex-mediated sodium iodide symporter (NIS) gene delivery. In the present study, we used novel polyplexes based on linear polyethylenimine (LPEI), polyethylene glycol (PEG), and the synthetic peptide GE11 as an epidermal growth factor receptor (EGFR)-specific ligand to target a NIS-expressing plasmid to hepatocellular carcinoma (HCC) (HuH7). Incubation of HuH7 cells with LPEI-PEG-GE11/NIS polyplexes resulted in a 22-fold increase in iodide uptake, which was confirmed in other cancer cell lines correlating well with EGFR expression levels. Using (123)I-scintigraphy and ex vivo γ-counting, HuH7 xenografts accumulated 6.5-9% injected dose per gram (ID/g) (123)I, resulting in a tumor-absorbed dose of 47 mGray/Megabecquerel (mGy/MBq) (131)Iodide ((131)I) after intravenous (i.v.) application of LPEI-PEG-GE11/NIS. No iodide uptake was observed in other tissues. After pretreatment with the EGFR-specific antibody cetuximab, tumoral iodide uptake was markedly reduced confirming the specificity of EGFR-targeted polyplexes. After three or four cycles of polyplex/(131)I application, a significant delay in tumor growth was observed associated with prolonged survival. These results demonstrate that systemic NIS gene transfer using polyplexes coupled with an EGFR-targeting ligand is capable of inducing tumor-specific iodide uptake, which represents a promising innovative strategy for systemic NIS gene therapy in metastatic cancers.


Subject(s)
ErbB Receptors/genetics , Genetic Therapy/methods , Iodine Radioisotopes/therapeutic use , Liver Neoplasms/therapy , Symporters/genetics , Cell Line, Tumor , ErbB Receptors/metabolism , Humans , Liver Neoplasms/radiotherapy , Polyethylene Glycols/chemistry , Polyethyleneimine/chemistry , Polymerase Chain Reaction , Polymers/administration & dosage , Polymers/chemistry
9.
Breast Cancer Res Treat ; 125(2): 377-86, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20300827

ABSTRACT

The sodium iodide symporter (NIS) mediates the active iodide uptake in the thyroid gland as well as lactating breast tissue. Recently, we reported significant stimulation of all-trans retinoic acid (atRA)-induced NIS expression in the estrogen-receptor positive human breast cancer cell line MCF-7 by dexamethasone (Dex) in vitro and in vivo, which might offer the potential to image and treat breast cancer with radioiodine. In this study, based on its known interaction with the pregnane-X-receptor (PXR) forming a heterodimer with the retinoid-X-receptor (RXR), we examined the effect of carbamazepine (CBZ), a potent activator of PXR, on atRA-induced NIS expression and therapeutic efficacy of (131)I in MCF-7 cells. For this purpose, functional NIS expression in MCF-7 cells was examined by iodide uptake assay, quantitative real-time PCR as well as Western blot analysis, followed by investigation of (131)I cytotoxicity in vitro after incubation with CBZ (4, 25, 100 µM) in the presence of atRA (1 µM) with or without Dex (100 nM). Incubation with CBZ stimulated atRA-induced iodide accumulation up to twofold in a concentration-dependent manner, while atRA/Dex-stimulated iodide uptake was further stimulated up to 1.5-fold by additional CBZ treatment based on significantly increased NIS mRNA and protein levels. This stimulatory effect of CBZ was shown to be dependent on the PI3K-Akt pathway without involvement of mTOR. In contrast, treatment with CBZ alone had no effect on functional NIS expression. Moreover, selective cytotoxicity of (131)I was significantly increased from approximately 20% in MCF-7 cells treated with atRA alone to 50% after treatment with CBZ in the presence of atRA, which was further enhanced to 90% after combined treatment with atRA/Dex/CBZ. In conclusion, CBZ represents another potent stimulator of atRA-induced functional NIS expression resulting in an enhanced selective killing effect of (131)I in MCF-7 breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Carbamazepine/pharmacology , Iodine Radioisotopes/pharmacology , Symporters/genetics , Symporters/metabolism , Tretinoin/pharmacology , Blotting, Western , Breast Neoplasms/genetics , Carbamazepine/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Dexamethasone/pharmacology , Female , Humans , Iodine Radioisotopes/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Polymerase Chain Reaction , Proto-Oncogene Proteins c-akt/metabolism , Retinoid X Receptors/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Stem Cell Assay
10.
Clin Cancer Res ; 15(19): 6079-86, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19789324

ABSTRACT

PURPOSE: We recently reported the significant therapeutic efficacy of radioiodine therapy in various tumor mouse models following transcriptionally targeted sodium iodide symporter (NIS) gene transfer. These studies showed the high potential of NIS as a novel diagnostic and therapeutic gene for the treatment of extrathyroidal tumors. As a next crucial step towards clinical application of NIS-mediated radionuclide therapy we aim at systemic delivery of the NIS gene to target extrathyroidal tumors even in the metastatic stage. EXPERIMENTAL DESIGN: In the current study, we used synthetic polymeric vectors based on pseudodendritic oligoamines with high intrinsic tumor affinity (G2-HD-OEI) to target a NIS-expressing plasmid (CMV-NIS-pcDNA3) to neuroblastoma (Neuro2A) cells. RESULTS: Incubation with NIS-containing polyplexes (G2-HD-OEI/NIS) resulted in a 51-fold increase in perchlorate-sensitive iodide uptake activity in Neuro2A cells in vitro. Through (123)I-scintigraphy and ex vivo gamma counting Neuro2A tumors in syngeneic A/J mice were shown to accumulate 8% to 13% ID/g (123)I with a biological half-life of 13 hours, resulting in a tumor-absorbed dose of 247 mGy/MBq (131)I after i.v. application of G2-HD-OEI/NIS. Nontarget organs, including liver, lung, kidneys, and spleen revealed no significant iodide uptake. Moreover, two cycles of systemic NIS gene transfer followed by (131)I application (55.5 MBq) resulted in a significant delay in tumor growth associated with markedly improved survival. CONCLUSIONS: In conclusion, our data clearly show the high potential of novel pseudodendritic polymers for tumor-specific NIS gene delivery after systemic application, opening the prospect of targeted NIS-mediated radionuclide therapy of nonthyroidal tumors even in metastatic disease.


Subject(s)
Genetic Therapy/methods , Iodine Radioisotopes/therapeutic use , Neuroblastoma/therapy , Radiotherapy , Symporters/genetics , Animals , Biological Availability , Combined Modality Therapy , Dendrimers/administration & dosage , Dendrimers/pharmacokinetics , Gene Transfer Techniques , Iodine Radioisotopes/pharmacokinetics , Male , Mice , Neoplasm Transplantation , Neuroblastoma/genetics , Neuroblastoma/metabolism , Neuroblastoma/pathology , Radiotherapy/methods , Radiotherapy, Adjuvant , Symporters/administration & dosage , Symporters/metabolism , Time Factors , Tumor Burden , Tumor Cells, Cultured
11.
Eur J Nucl Med Mol Imaging ; 35(7): 1272-81, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18404268

ABSTRACT

PURPOSE: We reported recently the induction of selective iodide uptake in prostate cancer cells (LNCaP) by prostate-specific antigen (PSA) promoter-directed sodium iodide symporter (NIS) expression that allowed a significant therapeutic effect of (131)I. In the current study, we studied the potential of the high-energy alpha-emitter (211)At, also transported by NIS, as an alternative radionuclide after NIS gene transfer in tumors with limited therapeutic efficacy of (131)I due to rapid iodide efflux. METHODS: We investigated uptake and therapeutic efficacy of (211)At in LNCaP cells stably expressing NIS under the control of the PSA promoter (NP-1) in vitro and in vivo. RESULTS: NP-1 cells concentrated (211)At in a perchlorate-sensitive manner, which allowed a dramatic therapeutic effect in vitro. After intraperitoneal injection of (211)At (1 MBq), NP-1 tumors accumulated approximately 16% ID/g (211)At (effective half-life 4.6 h), which resulted in a tumor-absorbed dose of 1,580+/-345 mGy/MBq and a significant tumor volume reduction of up to 82+/-19%, while control tumors continued their growth exponentially. CONCLUSIONS: A significant therapeutic effect of (211)At has been demonstrated in prostate cancer after PSA promoter-directed NIS gene transfer in vitro and in vivo suggesting a potential role for (211)At as an attractive alternative radioisotope for NIS-targeted radionuclide therapy, in particular in smaller tumors with limited radionuclide retention time.


Subject(s)
Astatine/therapeutic use , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Symporters/metabolism , Animals , Astatine/administration & dosage , Astatine/pharmacokinetics , Cell Line, Tumor , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Prostatic Neoplasms/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Symporters/genetics , Transfection , Transplantation, Heterologous , Tumor Stem Cell Assay
12.
Circulation ; 117(17): 2232-40, 2008 Apr 29.
Article in English | MEDLINE | ID: mdl-18427126

ABSTRACT

BACKGROUND: Prolonged myocardial ischemia results in cardiomyocyte loss despite successful revascularization. We have reported that retrograde application of embryonic endothelial progenitor cells (eEPCs) provides rapid paracrine protection against ischemia-reperfusion injury. Here, we investigated the role of thymosin beta4 (Tbeta4) as a mediator of eEPC-mediated cardioprotection. METHODS AND RESULTS: In vitro, neonatal rat cardiomyocytes were subjected to hypoxia-reoxygenation in the absence or presence of eEPCs with or without Tbeta4 short hairpin RNA (shRNA) transfection. In vivo, pigs (n=9 per group) underwent percutaneous left anterior descending artery occlusion for 60 minutes on day 1. After 55 minutes of ischemia, control eEPCs (5x10(6) cells) or cells transfected with Tbeta4 shRNA when indicated or 15 mg Tbeta4 alone were retroinfused into the anterior interventricular vein. Segmental endocardial shortening in the infarct zone at 150-bpm atrial pacing, infarct size (triphenyl tetrazolium chloride viability and methylene blue exclusion), and inflammatory cell influx (myeloperoxidase activity) were determined 24 hours later. Survival of neonatal rat cardiomyocytes increased from 32+/-4% to 90+/-2% after eEPC application, an effect sensitive to shRNA transfection compared with Tbeta4 (45+/-7%). In vivo, infarct size decreased with eEPC application (38+/-4% versus 54+/-4% of area at risk; P<0.01), an effect abolished by Tbeta4 shRNA (62+/-3%). Segmental subendocardial shortening improved after eEPC treatment (22+/-3% versus -3+/-4% of control area) unless Tbeta4 shRNA was transfected (-6+/-4%). Retroinfusion of Tbeta4 mimicked eEPC application (infarct size, 37+/-3%; segmental endocardial shortening, 34+/-7%). Myeloperoxidase activity (3323+/-388 U/mg in controls) was decreased by eEPCs (1996+/-546 U/mg) or Tbeta4 alone (1455+/-197 U/mg) but not Tbeta4 shRNA-treated eEPCs (5449+/-829 U/mg). CONCLUSIONS: Our findings show that short-term cardioprotection derived by regional application of eEPCs can be attributed, at least in part, to Tbeta4.


Subject(s)
Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/therapy , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Thymosin/genetics , Animals , Cell Adhesion/immunology , Cell Survival/physiology , Cells, Cultured , Embryonic Stem Cells/transplantation , Endothelial Cells/cytology , Endothelial Cells/physiology , Genetic Therapy/methods , Leukocytes/cytology , Mice , Mice, Transgenic , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/metabolism , Paracrine Communication/physiology , Rats , Stem Cell Transplantation , Swine , Thymosin/metabolism , Transfection
13.
Breast Cancer Res Treat ; 109(2): 263-72, 2008 May.
Article in English | MEDLINE | ID: mdl-17636401

ABSTRACT

CONTEXT: The sodium iodide symporter (NIS) mediates iodide uptake in the thyroid gland as well as in lactating breast, and is also expressed in the majority of breast cancers. Recently, we have reported stimulation of all-trans retinoic acid (atRA)-induced NIS expression in the human breast cancer cell line MCF-7 by dexamethasone (Dex), resulting in an enhanced therapeutic effect of (131)I in vitro. OBJECTIVE: In the current study we examined the efficacy of Dex stimulation of atRA-induced NIS expression in vivo in MCF-7 xenotransplants in nude mice. DESIGN: After systemic treatment with atRA alone or in combination with Dex, iodide accumulation in the tumors was assessed by gamma camera imaging and gamma counter analysis. In addition, NIS expression was examined on RNA and protein level by RT-PCR and immunohistochemistry, respectively. RESULTS: Using gamma camera imaging after intraperitoneal injection of 18.5 MBq (123)I, no iodide accumulation was detected in tumors of untreated mice or mice treated with atRA only. After combined treatment with atRA/Dex significant (123)I accumulation was detected in MCF-7 xenografts, which, by ex vivo gamma counting revealed a 3.3-fold increase in iodide accumulation as compared to control tumors. Surprisingly, in a subset of mice treated with atRA or atRA/Dex iodide accumulation was also detected in the normal mammary glands. In a normal human mammary epithelial cell line HB-2, however, no functional NIS expression was induced after treatment with atRA and/or Dex in vitro. Further, NIS mRNA and protein expression was detected in atRA/Dex treated MCF-7 tumors by RT-PCR and immunohistochemistry, respectively. CONCLUSION: Treatment with Dex in the presence of atRA is able to induce significant amounts of iodide accumulation in breast cancer xenotransplants in vivo due to stimulation of functional NIS protein expression, which opens exciting perspectives for a possible diagnostic and therapeutic role of radioiodine in the treatment of breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Symporters/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Dexamethasone/administration & dosage , Humans , Immunohistochemistry , Iodides/analysis , Mice , Mice, Nude , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Symporters/drug effects , Tretinoin/administration & dosage , Xenograft Model Antitumor Assays
14.
J Clin Endocrinol Metab ; 92(11): 4451-8, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17698909

ABSTRACT

CONTEXT: We reported recently the induction of iodide accumulation in prostate cancer cells (LNCaP) by prostate-specific antigen promoter-directed sodium iodide symporter (NIS) expression that allowed a significant therapeutic effect of (131)iodine ((131)I). These data demonstrated the potential of the NIS gene as a novel therapeutic gene, although in some extrathyroidal tumors, therapeutic efficacy may be limited by rapid iodide efflux due to a lack of iodide organification. OBJECTIVE: In the current study, we therefore studied the potential of (188)rhenium ((188)Re), as an alternative radionuclide, also transported by NIS, with a shorter half-life and higher energy beta-particles than (131)I. RESULTS: NIS-transfected LNCaP cells (NP-1) concentrated 8% of the total applied activity of (188)Re as compared with 16% of (125)I, which was sufficient for a therapeutic effect in an in vitro clonogenic assay. gamma-Camera imaging of NP-1 cell xenografts in nude mice revealed accumulation of 8-16% injected dose (ID)/g (188)Re (biological half-life 12.9 h), which resulted in a 4.7-fold increased tumor absorbed dose (450 mGy/MBq) for (188)Re as compared with (131)I. After application of 55.5 MBq (131)I or (188)Re, smaller tumors showed a similar average volume reduction of 86%, whereas in larger tumors volume reduction was significantly increased from 73% after (131)I treatment to 85% after application of (188)Re. CONCLUSION: Although in smaller prostate cancer xenografts both radionuclides seemed to be equally effective after prostate-specific antigen promoter-mediated NIS gene delivery, a superior therapeutic effect has been demonstrated for (188)Re in larger tumors.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy , Rhenium/therapeutic use , Symporters/genetics , Adult , Animals , Cell Line, Tumor , Cell Survival/genetics , Fluorescent Antibody Technique, Indirect , Half-Life , Humans , Iodine Radioisotopes/therapeutic use , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prostatic Neoplasms/genetics , Radioisotopes/pharmacokinetics , Radioisotopes/therapeutic use , Rhenium/pharmacokinetics , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
15.
Carcinogenesis ; 28(3): 595-610, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17056607

ABSTRACT

Interactions between cancer cells and the tissue microenvironment play an essential role in controlling tumor development and progression. Here, we report that stromal modulation induced by a biodegradable meshwork (Hyalograft 3D) inhibited tumor vascularization and invasion of the locally invasive low-grade malignant human HaCaT-ras II-4 keratinocytes in a surface xenotransplantation assay. The scaffold caused formation of an active granulation tissue that shifted to a fibrotic-type connective tissue with accumulation of myofibroblasts and collagen bundles. Most importantly, in transplants with scaffolds, the epithelial-stromal border was normalized developing an ultrastructurally complete basement membrane (BM) including hemidesmosomes. The observed reversion of the tumor phenotype was not due to decreased tumor cell proliferation but correlated with (i) normalization of epidermal differentiation, (ii) condensation of extracellular matrix (ECM) and (iii) reduction of peritumoral protease activity Furthermore, inhibited invasion was paralleled by eliminated tumor vascularization. This was substantiated by a diminished endothelial VEGF-receptor (VEGFR) expression and, in turn, by a concomitant increase in the ECM components thrombospondin-1 (TSP-1) and endostatin, known to impair angiogenesis. Even in transplants of the metastatic high-grade malignant HaCaT-ras A-5RT3 keratinocytes the anti-invasive effect of the scaffold-modulated stroma prevailed. Tumor vascularization and invasion was reduced and the epithelial tissue partially normalized including formation of stretches of BM. This clearly demonstrates that the scaffold-modulated connective tissue not only blocks tumor invasion but reverts the tumor phenotype. These novel findings underline the controlling function of tumor stroma and open new strategies of cancer therapy by targeting tumor stroma elements.


Subject(s)
Carcinoma, Squamous Cell/genetics , Hyaluronic Acid/therapeutic use , Neovascularization, Pathologic/prevention & control , Skin Neoplasms/genetics , Animals , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Humans , Hyaluronic Acid/analogs & derivatives , Keratinocytes , Mice , Mice, Nude , Neoplasm Invasiveness , Phenotype , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Transplantation, Heterologous
16.
J Investig Dermatol Symp Proc ; 11(1): 93-105, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17069016

ABSTRACT

Epidermal homeostasis is understood as the maintenance of epidermal tissue structure and function by a fine tuned regulatory mechanism balancing proliferation and cell loss by desquamation and apoptosis. The lack of appropriate experimental models has largely prevented a better understanding of the regulatory mechanisms controlling epidermal tissue homeostasis in human skin. Keratinocyte culture studies had revealed a strict dependency of regular epidermal differentiation on dermal interactions only accomplishable in three-dimensional skin models. As major drawbacks, conventional models, employing collagen hydrogels as dermal equivalents (DEs) exhibit a rather poor stability and limited lifespan. Here, we present an improved stabilized in vitro-model for long-term growth and differentiation of keratinocytes providing the basis for tissue homeostasis. Keratinocytes were grown on DEs reinforced by modified hyaluronic acid fibers (Hyalograft-3D) and colonized with skin fibroblasts, producing genuine dermis-type matrix. These skin equivalents (SEs) develop superior epidermal architecture with regular differentiation and ultrastructure. Critical aspects of differentiation, still unbalanced in early stages, are renormalized, most strikingly the coexpression of keratins K1/K10, downregulation of regeneration-associated keratins (K16), and restriction of K15 to the basal layer. The strict localization of integrins to basal cells underlining restored tissue polarity, the drop of keratinocyte growth rates towards physiological levels and the rapid formation of a mature basement membrane with abundant anchoring fibrils are altogether features fulfilling the criteria of tissue homeostasis. Therefore, these scaffold-based SEs not only allow for studying homeostasis control but also for the first time provide proper experimental conditions for establishing a stem cell niche in vitro.


Subject(s)
Epidermal Cells , Skin, Artificial , Tissue Engineering , Adult , Basement Membrane/chemistry , Cell Differentiation , Cell Proliferation , Cells, Cultured , Epidermis/physiology , Epidermis/ultrastructure , Extracellular Matrix Proteins/biosynthesis , Hemostasis , Humans , Keratinocytes/physiology , Keratins/analysis
17.
Eur J Cell Biol ; 83(11-12): 631-45, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15679108

ABSTRACT

Besides medical application as composite skin grafts, in vitro constructed skin equivalents (SEs) or organotypic co-cultures represent valuable tools for cutaneous biology. Major drawbacks of conventional models, employing collagen hydrogels as dermal equivalents (DEs), are a rather poor stability and limited life span, restricting studies to early phases of skin regeneration. Here we present an improved stabilised in vitro model actually providing the basis for skin-like homeostasis. Keratinocytes were grown on dermal equivalents (DEs) reinforced by modified hyaluronic acid fibres (Hyalograft-3D) and colonised with skin fibroblasts, producing genuine dermis-type matrix. These SEs developed a superior epidermal architecture with regular differentiation and ultrastructure, which occurred also faster than in SEs based on collagen-DEs. Critical aspects of differentiation, still unbalanced in early stages, were perfectly re-normalised, most strikingly the co-expression of keratins K1/K10 and downregulation of regeneration-associated keratins such as K16. The restriction of integrin and K15 distribution as well as keratinocyte proliferation to the basal layer underlined the restored tissue polarity, while the drop of growth rates towards physiological levels implied finally accomplishment of homeostasis. This correlated to faster basement membrane (BM) formation and ultrastructurally defined dermo-epidermal junction including abundant anchoring fibrils for strong tissue connection. Whereas the fibroblasts in the scaffold initially secreted a typical provisional regenerative matrix (fibronectin, tenascin), with time collagens of mature dermis (type I and III) were accumulating giving rise to an in vivo-like matrix with regularly organised bundles of striated collagen fibrils. In contrast to the more catabolic state in conventional DEs, the de novo reconstruction of genuine dermal tissue seemed to be a key element for maintaining prolonged normal keratinocyte proliferation (followed up to 8 wks), fulfilling the criteria of tissue-homeostasis, and possibly providing a stem cell niche.


Subject(s)
Dermis/physiology , Epidermis/growth & development , Extracellular Matrix Proteins/biosynthesis , Fibroblasts/physiology , Morphogenesis/physiology , Tissue Engineering/methods , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques , Epidermal Cells , Epidermis/ultrastructure , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Humans , Integrins/metabolism , Organ Culture Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...